Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Eur J Drug Metab Pharmacokinet ; 49(3): 343-353, 2024 May.
Article in English | MEDLINE | ID: mdl-38472634

ABSTRACT

BACKGROUND AND OBJECTIVE: In vitro glucuronidation of 17ß-estradiol (estradiol) is often performed to assess the role of uridine 5'-diphospho-glucuronosyltransferase 1A1 (UGT1A1) in xenobiotic/drug metabolism. The objective of this study was to determine the effects of four commonly used organic solvents [i.e., dimethyl sulfoxide (DMSO), methanol, ethanol, and acetonitrile] on the glucuronidation kinetics of estradiol, which can be glucuronidated at C3 and C17 positions. METHODS: The impacts of organic solvents on estradiol glucuronidation were determined by using expressed UGT enzymes and liver microsomes from both human and animals. RESULTS: In human liver microsomes (HLM), methanol, ethanol, and acetonitrile significantly altered estradiol glucuronidation kinetics with increased Vmax (up to 2.6-fold) and CLmax (up to 2.8-fold) values. Altered estradiol glucuronidation in HLM was deduced to be attributed to the enhanced metabolic activities of UGT1A1 and UGT2B7, whose activities differ at the two glucuronidation positions. The effects of organic solvents on estradiol glucuronidation were glucuronidation position-, isozyme-, and solvent-specific. Furthermore, both ethanol and acetonitrile have a greater tendency to modify the glucuronidation activity of estradiol in animal liver microsomes. CONCLUSION: Organic solvents such as methanol, ethanol, and acetonitrile showed great potential in adjusting the glucuronidation of estradiol. DMSO is the most suitable solvent due to its minimal influence on estradiol glucuronidation. Researchers should be cautious in selecting appropriate solvents to get accurate results when assessing the metabolism of a new chemical entity.


Subject(s)
Dimethyl Sulfoxide , Estradiol , Ethanol , Glucuronides , Glucuronosyltransferase , Microsomes, Liver , Solvents , Microsomes, Liver/metabolism , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Estradiol/metabolism , Estradiol/pharmacology , Glucuronosyltransferase/metabolism , Humans , Solvents/pharmacology , Animals , Kinetics , Ethanol/metabolism , Ethanol/pharmacology , Glucuronides/metabolism , Dimethyl Sulfoxide/pharmacology , Methanol/pharmacology , Methanol/metabolism , Acetonitriles/pharmacology , Acetonitriles/metabolism
2.
Xenobiotica ; 53(3): 215-222, 2023 Mar.
Article in English | MEDLINE | ID: mdl-37039301

ABSTRACT

BCRP (breast cancer resistance protein) is a crucial efflux transporter involved in the regulation of the pharmacokinetics and pharmacodynamics of a wide range of drugs. Herein, we aimed to investigate a potential role for the nuclear receptor REV-ERBα in the regulation of BCRP expression and sulfasalazine (a BCRP probe substrate) pharmacokinetics.Regulation of BCRP expression by REV-ERBα was assessed using Rev-erbα-/- mice and AML12 and CT26 cells. Pharmacokinetic analysis was performed with Rev-erbα-/- and wild-type mice after sulfasalazine administration.We found that the expression levels of BCRP mRNA and protein were downregulated in the liver and small intestine of Rev-erbα-dificient mice. In line with this, Rev-erbα ablation increased the systemic exposures of oral sulfasalazine.Positive regulation of BCRP expression and function by REV-ERBα was furtherly confirmed in AML12 and CT26 cells. Moreover, indirect regulation of Bcrp expression by REV-ERBα was potentially mediated by a negative transcription factor DEC2, which is a downstream target of REV-ERBα.In conclusion, REV-ERBα positively regulates BCRP expression in mice, thereby affecting sulfasalazine pharmacokinetics.


Subject(s)
Neoplasm Proteins , Sulfasalazine , Mice , Animals , Sulfasalazine/pharmacology , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Neoplasm Proteins/genetics , Gene Expression Regulation , Receptors, Cytoplasmic and Nuclear
3.
Biochem Pharmacol ; 205: 115286, 2022 11.
Article in English | MEDLINE | ID: mdl-36216079

ABSTRACT

Delirium is a common and serious neuropsychiatric syndrome characterized with acute cognitive and attentional deficits, however, the effective therapies are lacking. Here, using mouse models of delirium, we investigated the effects of tangeretin (TAN, a natural flavonoid) on cognitive impairment by assessing object preference with novel object recognition (NOR) test and spontaneous alternation with Y maze test. We found that TAN, as a RORα/γ agonist, prevented cognitive decline in delirious mice as evidenced by a normal novel object preference and increased spontaneous alternation. This was accompanied by decreased expression of ERK1/2, TNFα and IL-1ß as well as diminished microglial activation in delirious mice. The protective effect of TAN on delirium was mainly attributed to increased hippocampal E4BP4 expression (a known target of RORs and a regulator of cognition in delirium through modulating the ERK1/2 cascade and microglial activation) via activation of RORα/γ. In addition, TAN treatment modulated the expression of RORα/γ target genes (such as E4bp4 and Bmal1) and inhibited the expression of TNFα and IL-1ß in lipopolysaccharide (LPS)-stimulated cells, supporting a beneficial effect of TAN on delirium. In conclusion, TAN is identified as a RORα/γ agonist which promotes E4BP4 expression to prevent cognitive decline in delirious mice. Our findings may have implications for drug development of TAN for prevention and treatment of various diseases associated with cognitive deficiency.


Subject(s)
Cognitive Dysfunction , Delirium , Flavones , Animals , Mice , ARNTL Transcription Factors , Cognition , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/prevention & control , Cognitive Dysfunction/metabolism , Flavones/pharmacology , Lipopolysaccharides , Tumor Necrosis Factor-alpha/genetics
4.
Cell Rep ; 38(10): 110506, 2022 03 08.
Article in English | MEDLINE | ID: mdl-35263590

ABSTRACT

For survival, animals encode prominent events in complex environments, which modulates their defense behavior. Here, we design a paradigm that assesses how a mild aversive cue (i.e., mild air puff) interacts with sound-evoked flight behavior in mice. We find that air puffing facilitates sound-evoked flight behavior by enhancing the auditory responses of auditory cortical neurons. We then find that the anterior part of the anterior cingulate cortex (ACC) encodes the valence of air puffing and modulates the auditory cortex through anatomical examination, physiological recordings, and optogenetic/chemogenetic manipulations. Activating ACC projections to the auditory cortex simulates the facilitating effect of air puffing, whereas inhibiting the ACC or its projections to the auditory cortex neutralizes this facilitating effect. These findings show that the ACC regulates sound-evoked flight behavior by potentiating neuronal responses in the auditory cortex.


Subject(s)
Auditory Cortex , Gyrus Cinguli , Animals , Auditory Cortex/physiology , Mice , Neurons/physiology , Optogenetics
5.
Curr Drug Metab ; 23(1): 2-7, 2022.
Article in English | MEDLINE | ID: mdl-34994324

ABSTRACT

Dosing time-dependency of pharmacokinetics (or chronopharmacokinetics) has been long recognized. Studies in recent years have revealed that daily rhythmicity in expression of drug-metabolizing enzymes and transporters (DMETs) are key factors determining chronopharmacokinetics. In this article, we briefly summarize current knowledge with respect to circadian mechanisms of DMETs and discuss how rhythmic DMETs are translated to drug chronoeffects. More importantly, we present our perspectives on pharmacokinetics-based chronotherapy.


Subject(s)
Circadian Clocks , Chronopharmacokinetics , Chronotherapy , Circadian Rhythm , Humans
6.
Front Pharmacol ; 13: 1088294, 2022.
Article in English | MEDLINE | ID: mdl-36618934

ABSTRACT

Puerariae lobatae radix (PLR) is a wildly used herbal medicine. Here we aimed to assess the PLR efficacy against UVB (ultraviolet-B)-induced skin aging and to determine the mechanisms thereof. We found a significant protective effect of PLR (topical application) on UVB-induced skin aging in mice, as evidenced by reduced skin wrinkles, epidermal thickness, and MDA (malondialdehyde) content as well as increased levels of HYP (hydroxyproline) and SOD (superoxide dismutase) in the skin. In the meantime, Mmp-1, p21 and p53 levels were decreased in the skin of PLR-treated mice. Anti-aging effects of PLR were also confirmed in L929 cells. Furthermore, PLR up-regulated skin expression of BMAL1, which is a known regulator of aging by promoting Nrf2 and antioxidant enzymes. Consistently, Nrf2 and several genes (i.e., Prdx6, Sod1, and Sod2) encoding antioxidant enzymes in the skin were increased in PLR-treated mice. Moreover, based on Gal4 chimeric assay, Bmal1 reporter gene and expression assays, we identified PLR as an antagonist of REV-ERBα that can increase Bmal1 expression. Intriguingly, loss of Rev-erbα protected mice against UVB-induced skin aging and abrogated the protective effect of PLR. In conclusion, PLR acts as an antagonist of REV-ERBα and promotes the expression of BMAL1 to protect against skin aging in mice.

7.
Front Pharmacol ; 12: 764124, 2021.
Article in English | MEDLINE | ID: mdl-34887762

ABSTRACT

CYP2B10 is responsible for metabolism and detoxification of many clinical drugs. Here, we aimed to investigate a potential role of Period 2 (PER2) in regulating expression of hepatic CYP2B10. Regulatory effects of PER2 on hepatic expression of CYP2B10 and other enzymes were determined using Per2-deficient mice with exons 4-6 deleted (named Per2 Del4-6 mice). In vitro and in vivo metabolic activities of CYP2B10 were probed using cyclophosphamide (CPA) as a specific substrate. Regulatory mechanism was investigated using luciferase reporter assays. Genotyping and Western blotting demonstrated loss of wild-type Per2 transcript and markedly reduced PER2 protein in Per2 Del4-6 mice. Hepatic expression of a plenty of drug-metabolizing genes (including Cyp2a4/2a5, Cyp2b10, Ugt1a1, Ugt1a9, Ugt2b36, Sult1a1 and Sult1e1) were altered (and majority were down-regulated) in Per2 Del4-6 mice. Of note, Cyp2b10, Ugt1a9 and Sult1a1 were three genes considerably affected with reduced expression. Decreased expression of CYP2B10 was translated to reduced metabolism and altered pharmacokinetics of CPA as well as attenuated CPA hepatotoxicity in Per2 Del4-6 mice. Positive regulation of CYP2B10 by PER2 was further confirmed in both Hepa-1c1c7 and AML-12 cells. Based on luciferase reporter assays, it was shown that PER2 regulated Cyp2b10 transcription in a REV-ERBα-dependent manner. REV-ERBα was negatively regulated by PER2 (increased REV-ERBα expression in Per2 Del4-6 mice) and itself was also a repressor of CYP2B10. In conclusion, PER2 positively regulates CYP2B10 expression and activity in mouse liver through inhibiting its repressor REV-ERBα.

8.
Biochem Pharmacol ; 193: 114807, 2021 11.
Article in English | MEDLINE | ID: mdl-34673015

ABSTRACT

The molecular events underlying aristolochic acid (AA) nephropathy are poorly understood, and specific therapies for treatment of AA nephropathy are still lacking. Here we aimed to investigate a potential role of REV-ERBα and ferroptosis in renal injury induced by aristolochic acid I (AAI), a typical AA. The regulatory effects of REV-ERBα on AAI-induced renal injury were determined using kidney-specific Rev-erbα knockout mice. Ferroptosis was assessed based on measurements of iron, GSH, and GPX4. Targeted antagonism of REV-ERBα to alleviate AAI-induced renal injury and ferroptosis was assessed using the small molecule antagonist SR8278. mRNAs and proteins were quantified by qPCR and Western blotting, respectively. We first showed that REV-ERBα was upregulated and its target BMAL1 was downregulated in the kidney of mice with AAI nephropathy. Upregulation of REV-ERBα protein was confirmed in aristolactam I (ALI, a nephrotoxic metabolite of AAI)-treated mRTECs. We also observed enhanced ferroptosis (known to be regulated by REV-ERBα) in mice with AAI nephropathy and in ALI-treated mRTECs. Kidney-specific knockout of Rev-erbα reduced the sensitivity of mice to AAI-induced ferroptosis and renal injury. Furthermore, knockdown of Rev-erbα by siRNA or SR8278 (a REV-ERBα antagonist) treatment attenuated ALI-induced ferroptosis in mRTECs. Moreover, REV-ERBα antagonism by SR8278 alleviated ferroptosis and renal injury caused by AAI in mice. In conclusion, we identify REV-ERBα as a regulator of AAI-induced renal injury via promoting ferroptosis. Targeting REV-ERBα may represent a promising approach for management of AAI nephropathy.


Subject(s)
Aristolochic Acids/toxicity , Ferroptosis/drug effects , Isoquinolines/pharmacology , Kidney Diseases/chemically induced , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , Thiophenes/pharmacology , Animals , Cell Line , Cell Survival/drug effects , Epithelial Cells/drug effects , Gene Expression Regulation/drug effects , Humans , Kidney Diseases/drug therapy , Kidney Tubules/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Structure , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics
9.
Nat Nanotechnol ; 16(10): 1150-1160, 2021 10.
Article in English | MEDLINE | ID: mdl-34354264

ABSTRACT

Although nanomaterials have shown promising biomedical application potential, incomplete understanding of their molecular interactions with biological systems prevents their inclusion into mainstream clinical applications. Here we show that black phosphorus (BP) nanomaterials directly affect the cell cycle's centrosome machinery. BP destabilizes mitotic centrosomes by attenuating the cohesion of pericentriolar material and consequently leads to centrosome fragmentation within mitosis. As a result, BP-treated cells exhibit multipolar spindles and mitotic delay, and ultimately undergo apoptosis. Mechanistically, BP compromises centrosome integrity by deactivating the centrosome kinase polo-like kinase 1 (PLK1). BP directly binds to PLK1, inducing its aggregation, decreasing its cytosolic mobility and eventually restricting its recruitment to centrosomes for activation. With this mechanism, BP nanomaterials show great anticancer potential in tumour xenografted mice. Together, our study reveals a molecular mechanism for the tumoricidal properties of BP and proposes a direction for biomedical application of nanomaterials by exploring their intrinsic bioactivities.


Subject(s)
Cell Cycle Proteins/genetics , Centrosome/drug effects , Nanostructures/chemistry , Neoplasms/drug therapy , Phosphorus/pharmacology , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Animals , Apoptosis/drug effects , Cell Cycle Proteins/antagonists & inhibitors , HeLa Cells , Heterografts , Humans , Mice , Mitosis/drug effects , Neoplasms/genetics , Neoplasms/pathology , Phosphorus/chemistry , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Polo-Like Kinase 1
10.
Front Pharmacol ; 12: 707844, 2021.
Article in English | MEDLINE | ID: mdl-34393786

ABSTRACT

Identifying drugs with dosing time-dependent effects (chronoeffects) and understanding the underlying mechanisms would help to improve drug treatment outcome. Here, we aimed to determine chronoeffects of the herbal medicines Puerariae radix (PR) and Coptidis rhizoma (CR), and investigate a potential role of REV-ERBα as a drug target in generating chronoeffects. The pharmacological effect of PR on hyperhomocysteinemia in mice was evaluated by measuring total homocysteine, triglyceride levels and lipid accumulation. PR dosed at ZT10 generated a stronger effect on hyperhomocysteinemia than drug dosed at ZT2. Furthermore, PR increased the expression levels of REV-ERBα target genes Bhmt, Cbs and Cth (encoding three key enzymes responsible for homocysteine catabolism), thereby alleviating hyperhomocysteinemia in mice. Moreover, CR attenuated chronic colitis in mice in a dosing time-dependent manner based on measurements of disease activity index, colon length, malondialdehyde/myeloperoxidase activities and IL-1ß/IL-6 levels. ZT10 dosing generated a stronger anti-colitis effect as compared to ZT2 dosing. This was accompanied by lower production of colonic inflammatory cytokines (i.e., Nlrp3, IL-1ß, IL-6, Tnf-α and Ccl2, REV-ERBα target genes) in colitis mice dosed at ZT10. The diurnal patterns of PR and CR effects were respectively consistent with those of puerarin (a main active constituent of PR, a REV-ERBα antagonist) and berberine (a main active constituent of CR, a REV-ERBα agonist). In addition, loss of Rev-erbα in mice abolished the dosing time-dependency in PR and CR effects. In conclusion, the therapeutic effects of PR and CR depend on dosing time in mice, which are probably attributed to diurnal expression of REV-ERBα as the drug target. Our findings have implications for improving therapeutic outcomes of herbal medicines with a chronotherapeutic approach.

11.
Xenobiotica ; 51(9): 1019-1028, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34311664

ABSTRACT

1. Retrorsine (RTS) is a pyrrolizidine alkaloid (distributed in many medicinal plants) that has significant hepatotoxicity. Here, we aimed to determine the daily variations in RTS hepatotoxicity (chronotoxicity) in mice, and to investigate the role of metabolism in generating RTS chronotoxicity.2. Acute toxicity and pharmacokinetic studies were performed with mice after RTS administration at different times of the day. Hepatotoxicity was assessed by measuring plasma ALT (alanine aminotransferase) and AST (aspartate aminotransferase) levels. mRNA and proteins were determined by qPCR and Western blotting, respectively. Time-dependent in vitro metabolism of RTS was assessed by using mouse liver microsomes.3. We found that RTS toxicity was more severe in the dark phase (zeitgeber time 14 or ZT14 and ZT18) than in the light phase (ZT2 and ZT6). This chronotoxicity was associated with a dosing time difference in the systemic exposures of RTS and a pyrrolic ester metabolite (a cause of hepatotoxicity, measured by the levels of pyrrole-GSH conjugate and pyrrole-protein adducts due to a high chemical reactivity). Moreover, the CYP3A11 (a major enzyme for RTS bioactivation) inhibitor ketoconazole decreased the production of pyrrole-GSH conjugate and abrogated diurnal rhythm in RTS metabolism. In addition, E4bp4 (a circadian regulator of Cyp3a11) ablation abolished the rhythm of CYP3A11 expression and abrogated the dosing time-dependency of RTS toxicity.4. In conclusion, RTS chronotoxicity in mice was attributed to time-varying hepatic metabolism regulated by the circadian clock. Our findings have implications for reducing pyrrolizidine alkaloid-induced toxicity via a chronotherapeutic approach.


Subject(s)
Circadian Clocks , Pyrrolizidine Alkaloids , Alanine Transaminase , Animals , Circadian Rhythm , Cytochrome P-450 CYP3A/genetics , Liver , Membrane Proteins , Mice , Pyrrolizidine Alkaloids/toxicity
12.
Biochem Biophys Res Commun ; 571: 167-173, 2021 09 24.
Article in English | MEDLINE | ID: mdl-34330060

ABSTRACT

Aurora kinase A (Aurora A) plays a critical role in regulating cell mitotic progression and has been considered as a promising drug target for cancer therapy. To develop a novel molecule targeting Aurora A with high selectivity and efficacy, we designed and synthesized a pyrrole-imidazole polyamide (PIP) Hoechst conjugate, PIP-Ht, targeting to a cell-cycle regulated DNA sequence locating at the promoter of human Aurora A gene (AURKA). PIP-Ht potently suppressed AURKA promoter activities, mRNA expression and protein level, induced tumor cell cycle delay and inhibited tumor cell proliferation in vitro. Furthermore, subcutaneous injection of PIP-Ht into mice bearing human cancer xenografts induced significant tumor growth suppression and cell apoptosis. Collectively, PIP-Ht exhibits the potential as an effective therapeutic candidate for the tumor treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Aurora Kinase A/antagonists & inhibitors , Imidazoles/pharmacology , Nylons/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrroles/pharmacology , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Aurora Kinase A/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Female , Humans , Imidazoles/chemistry , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Nylons/chemistry , Protein Kinase Inhibitors/chemistry , Pyrroles/chemistry , Tumor Cells, Cultured
13.
J Pharm Pharmacol ; 73(3): 398-409, 2021 Mar 06.
Article in English | MEDLINE | ID: mdl-33793874

ABSTRACT

OBJECTIVES: We aimed to determine the circadian responses of mice to Semen Strychni and to investigate the role of pharmacokinetics in generating chronotoxicity. METHODS: Total extract of Semen Strychni was administered by oral gavage to wild-type (WT) and Bmal1-/- (a circadian clock-deficient model) mice at different circadian time points for toxicity (including survival) and pharmacokinetic characterization. Nephrotoxicity and neurotoxicity were evaluated by measuring plasma creatinine and creatine kinase BB (CK-BB), respectively. Drug metabolism and transport assays were performed using liver/intestine microsomes and everted gut sacs, respectively. KEY FINDINGS: Semen Strychni nephrotoxicity and neurotoxicity as well as animal survival displayed significant circadian rhythms (the highest level of toxicity was observed at ZT18 and the lowest level at ZT2 to ZT6). According to pharmacokinetic experiments, herb dosing at ZT18 generated higher plasma concentrations (and systemic exposure) of strychnine and brucine (two toxic constituents) compared with ZT6 dosing. This was accompanied by reduced formation of both dihydroxystrychnine and strychnine glucuronide (two strychnine metabolites) at ZT18. Bmal1 ablation sensitized mice to Semen Strychni-induced toxicity (with increased levels of plasma creatinine and CK-BB) and abolished the time dependency of toxicity. Metabolism of Semen Strychni (strychnine and brucine) in the liver and intestine microsomes of WT mice was more extensive at ZT6 than at ZT18. These time differences in hepatic and intestinal metabolism were lost in Bmal1-/- mice. Additionally, the intestinal efflux transport of Semen Strychni (strychnine and brucine) was more extensive at ZT6 than ZT18 in WT mice. However, the time-varying transport difference was abolished in Bmal1-/- mice. CONCLUSIONS: Circadian responses of mice to Semen Strychni are associated with time-varying efflux transport and metabolism regulated by the circadian clock (Bmal1). Our findings may have implications for optimizing phytotherapy with Semen Strychni via timed delivery.


Subject(s)
ARNTL Transcription Factors/genetics , Circadian Rhythm/physiology , Plant Extracts/toxicity , Strychnos nux-vomica/chemistry , Animals , Biological Transport , Circadian Clocks/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microsomes/metabolism , Neurotoxicity Syndromes/etiology , Plant Extracts/administration & dosage , Plant Extracts/pharmacokinetics , Strychnine/analogs & derivatives , Strychnine/pharmacokinetics , Strychnine/toxicity , Time Factors
14.
Biochem Biophys Res Commun ; 547: 52-58, 2021 04 02.
Article in English | MEDLINE | ID: mdl-33592379

ABSTRACT

Natural products are useful tools for biological mechanism research and drug discovery. Due to the excellent tumor cell growth inhibitory profile and sub-nanomolar potency, Coibamide A (CA), an N-methyl-stabilized depsipeptide isolated from marine cyanobacterium, has been considered as a promising lead compound for cancer treatment. However, the molecular anti-cancer mechanism of the action of CA remains unclear. Here, we showed that CA treatment induced caspase-independent cell death in breast cancer cells. CA treatment also led to severe lysosome defects, which was ascribed to the impaired glycosylation of lysosome membrane protein LAMP1 and LAMP2. As a consequence, the autophagosome-lysosome fusion was blocked upon CA treatment. In addition, we presented evidence that this autophagy defect partially contributed to the CA treatment-induced tumor cell death. Together, our work uncovers a novel mechanism underlying the anti-cancer action of CA, which will promote its further application for cancer therapy.


Subject(s)
Autophagosomes/drug effects , Breast Neoplasms/drug therapy , Depsipeptides/pharmacology , Lysosomes/drug effects , Antineoplastic Agents/pharmacology , Autophagosomes/metabolism , Autophagy/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Lysosomes/metabolism , Signal Transduction
15.
J Med Chem ; 64(2): 991-1000, 2021 01 28.
Article in English | MEDLINE | ID: mdl-33417771

ABSTRACT

Coibamide A (1) is a highly N-methylated cyclodepsipeptide with low nanomolar antiproliferative activities against various cancer cell lines. In previous work, we discovered a simplified analogue, [MeAla3-MeAla6]-coibamide (1a), which exhibited the same inhibitory abilities as coibamide A. Herein, to reduce the whole-body toxicity and improve the solubility of 1a, two novel peptide-drug conjugates RGD-SS-CA (2) and RGD-VC-CA (3) were designed, synthesized, and evaluated. Composed of cyclodepsipeptide 1a, a tumor-homing RGD motif, and a conditionally labile linker, the conjugates are expected to release 1a tracelessly in specific tumor microenvironments. Compared with RGD-VC-CA (3), RGD-SS-CA (2) proved to be superior in in vitro drug release and cytotoxicity tests. Notably, intravenous injection of RGD-SS-CA (2) into mice-bearing human tumor xenografts induced significant tumor growth suppression with negligible toxicity. Therefore, as a novel prodrug of the coibamide A analogue, conjugate 2 has great potential for further exploration in cancer drug discovery.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Depsipeptides/chemical synthesis , Depsipeptides/pharmacology , Animals , Antineoplastic Agents/toxicity , Cell Line, Tumor , Cell Proliferation/drug effects , Depsipeptides/chemistry , Depsipeptides/toxicity , Drug Design , Humans , Male , Mice , Mice, Inbred BALB C , Prodrugs/pharmacology , Solubility , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays
16.
Chem Commun (Camb) ; 56(66): 9537-9540, 2020 Aug 19.
Article in English | MEDLINE | ID: mdl-32691026

ABSTRACT

A convenient and efficient strategy was developed for accessing chlorotoxin-derived bicyclic peptide-biomolecule conjugates by cyclizing fully-unprotected linear peptides with a designed tetrafunctional chemical linker. Among these peptides, bicycle-P3 bearing the N-terminal sequence of chlorotoxin shows high tumor selectivity and penetration ability, which is promising for treatment of gliomas.


Subject(s)
Brain Neoplasms/diagnostic imaging , Glioblastoma/diagnostic imaging , Optical Imaging/methods , Peptides, Cyclic/chemistry , Scorpion Venoms/chemistry , Amino Acid Sequence , Animals , Cell Line, Tumor , Fluorescent Dyes/chemistry , Humans , Mice , Transplantation, Heterologous
17.
J Pharm Pharmacol ; 72(12): 1854-1864, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32478421

ABSTRACT

OBJECTIVES: We aimed to determine the diurnal rhythm of Tripterygium wilfordii (TW) hepatotoxicity and to investigate a potential role of metabolism and pharmacokinetics in generating chronotoxicity. METHODS: Hepatotoxicity was determined based on assessment of liver injury after dosing mice with TW at different circadian time points. Circadian clock control of metabolism, pharmacokinetics and hepatotoxicity was investigated using Clock-deficient (Clock-/- ) mice. KEY FINDINGS: Hepatotoxicity of TW displayed a significant circadian rhythm (the highest level of toxicity was observed at ZT2 and the lowest level at ZT14). Pharmacokinetic experiments showed that oral gavage of TW at ZT2 generated higher plasma concentrations (and systemic exposure) of triptolide (a toxic constituent) compared with ZT14 dosing. This was accompanied by reduced formation of triptolide metabolites at ZT2. Loss of Clock gene sensitized mice to TW-induced hepatotoxicity and abolished the time-dependency of toxicity that was well correlated with altered metabolism and pharmacokinetics of triptolide. Loss of Clock gene also decreased Cyp3a11 expression in mouse liver and blunted its diurnal rhythm. CONCLUSIONS: Tripterygium wilfordii chronotoxicity was associated with diurnal variations in triptolide pharmacokinetics and circadian expression of hepatic Cyp3a11 regulated by circadian clock. Our findings may have implications for improving TW treatment outcome with a chronotherapeutic approach.


Subject(s)
CLOCK Proteins/metabolism , Chemical and Drug Induced Liver Injury/etiology , Circadian Rhythm/drug effects , Diterpenes/toxicity , Liver/drug effects , Phenanthrenes/toxicity , Plant Extracts/toxicity , Tripterygium/toxicity , Activation, Metabolic , Animals , CLOCK Proteins/genetics , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Cytochrome P-450 CYP3A/genetics , Cytochrome P-450 CYP3A/metabolism , Diterpenes/isolation & purification , Diterpenes/pharmacokinetics , Epoxy Compounds/isolation & purification , Epoxy Compounds/pharmacokinetics , Epoxy Compounds/toxicity , Liver/metabolism , Liver/pathology , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Phenanthrenes/isolation & purification , Phenanthrenes/pharmacokinetics , Plant Extracts/isolation & purification , Plant Extracts/pharmacokinetics , Toxicokinetics
18.
Drug Metab Dispos ; 48(5): 395-406, 2020 05.
Article in English | MEDLINE | ID: mdl-32114506

ABSTRACT

Dependence of drug metabolism on dosing time has long been recognized. However, only recently are the underlying mechanisms for circadian drug metabolism being clarified. Diurnal rhythmicity in expression of drug-metabolizing enzymes is believed to be a key factor determining circadian metabolism. Supporting the notion that biological rhythms are generated and maintained by the circadian clock, a number of diurnal enzymes are under the control of the circadian clock. In general, circadian clock genes generate and regulate diurnal rhythmicity in drug-metabolizing enzymes via transcriptional actions on one or two of three cis-elements (i.e., E-box, D-box, and Rev-erb response element or RAR-related orphan receptor response element). Additionally, cycling or clock-controlled nuclear receptors such as hepatocyte nuclear factor 4α and peroxisome proliferator-activated receptor γ are contributors to diurnal enzyme expression. These newly discovered mechanisms for each of the rhythmic enzymes are reviewed in this article. We also discuss how the rhythms of enzymes are translated to circadian pharmacokinetics and drug chronotoxicity, which has direct implications for chronotherapeutics. Our discussion is also extended to two diurnal transporters (P-glycoprotein and multidrug resistance-associated protein 2) that have an important role in drug absorption. Although the experimental evidence is lacking in metabolism-based chronoefficacy, circadian genes (e.g., Rev-erbα) as drug targets are shown to account for diurnal variability in drug efficacy. SIGNIFICANCE STATEMENT: Significant progress has been made in understanding the molecular mechanisms for generation of diurnal rhythmicity in drug-metabolizing enzymes. In this article, we review the newly discovered mechanisms for each of the rhythmic enzymes and discuss how the rhythms of enzymes are translated to circadian pharmacokinetics and drug chronotoxicity, which has direct implications for chronotherapeutics.


Subject(s)
Circadian Clocks/genetics , Drug Chronotherapy , Metabolic Clearance Rate/genetics , Multidrug Resistance-Associated Proteins/genetics , Animals , Humans , Models, Animal , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/metabolism , Response Elements , Transcriptional Activation , Treatment Outcome
19.
J Neurosci ; 40(10): 2025-2037, 2020 03 04.
Article in English | MEDLINE | ID: mdl-31980587

ABSTRACT

Plastic change in neuronal connectivity is the foundation of memory encoding. It is not clear whether the changes during anesthesia can alter subsequent behavior. Here, we demonstrated that in male rodents under anesthesia, a visual stimulus (VS) was associated with electrical stimulation of the auditory cortex or natural auditory stimulus in the presence of cholecystokinin (CCK), which guided the animals' behavior in a two-choice auditory task. Auditory neurons became responsive to the VS after the pairings. Moreover, high-frequency stimulation of axon terminals of entorhinal CCK neurons in the auditory cortex enabled LTP of the visual response in the auditory cortex. Such pairing during anesthesia also generated VS-induced freezing in an auditory fear conditioning task. Finally, we verified that direct inputs from the entorhinal CCK neurons and the visual cortex enabled the above neural plasticity in the auditory cortex. Our findings suggest that CCK-enabled visuoauditory association during anesthesia can be translated to the subsequent behavior action.SIGNIFICANCE STATEMENT Our study provides strong evidence for the hypothesis that cholecystokinin plays an essential role in the formation of cross-modal associative memory. Moreover, we demonstrated that an entorhinal-neocortical circuit underlies such neural plasticity, which will be helpful to understand the mechanisms of memory formation and retrieval in the brain.


Subject(s)
Cholecystokinin/metabolism , Entorhinal Cortex/physiology , Memory/physiology , Neural Pathways/physiology , Neuronal Plasticity/physiology , Acoustic Stimulation , Anesthesia , Animals , Association Learning/physiology , Auditory Cortex/physiology , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Photic Stimulation , Rats , Rats, Sprague-Dawley , Visual Cortex/physiology
20.
Chin J Nat Med ; 16(11): 871-880, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30502769

ABSTRACT

Poliumoside is representative of phenylethanoid glycosides, which are widely found in many plants. Poliumoside is also regarded as the main active component of Callicarpa kwangtungensis Chun (CK), though its oral bioavailability in rat is extremely low (0.69%) and its in vivo and in vitro metabolism has not yet been systematically investigated. In the present study, an ultra performance liquid chromatography/quadrupole time-of-flight mass spectrometry (UPLC/Q-TOF-MS) method was employed to identify the metabolites and investigate the metabolic pathways of poliumoside in rat after oral administration 1.5 g·kg-1 of poliumoside. As a result, a total of 34 metabolites (30 from urine, 17 from plasma, and 4 from bile) and 9 possible metabolic pathways (rearrangment, reduction, hydration, hydrolyzation, dehydration, methylation, hydroxylation, acetylation, and sulfation) were proposed in vivo. The main metabolite, acteoside, was quantified after incubated with rat intestinal bacteria in vitro. In conclusion, the present study systematically explored the metabolites of poliumoside in vivo and in vitro, proposing metabolic pathways that may be significant for further metabolic studies of poliumoside.


Subject(s)
Bacteria/metabolism , Bile/chemistry , Caffeic Acids/chemistry , Callicarpa/chemistry , Drugs, Chinese Herbal/chemistry , Glycosides/chemistry , Intestines/microbiology , Plasma/chemistry , Urine/chemistry , Administration, Oral , Animals , Caffeic Acids/administration & dosage , Caffeic Acids/blood , Caffeic Acids/urine , Chromatography, High Pressure Liquid , Drugs, Chinese Herbal/administration & dosage , Drugs, Chinese Herbal/metabolism , Glycosides/administration & dosage , Glycosides/blood , Glycosides/urine , Male , Mass Spectrometry/methods , Molecular Structure , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...